1
|
Oikonomou A, Valsecchi L, Quadri M, Watrin T, Scharov K, Procopio S, Tu JW, Vogt M, Savino AM, Silvestri D, Valsecchi MG, Biondi A, Borkhardt A, Bhatia S, Cazzaniga G, Fazio G, Bardini M, Palmi C. High-throughput screening as a drug repurposing strategy for poor outcome subgroups of pediatric B-cell precursor Acute Lymphoblastic Leukemia. Biochem Pharmacol 2023; 217:115809. [PMID: 37717691 DOI: 10.1016/j.bcp.2023.115809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Although a great cure rate has been achieved for pediatric BCP-ALL, approximately 15% of patients do not respond to conventional chemotherapy and experience disease relapse. A major effort to improve the cure rates by treatment intensification would result in an undesirable increase in treatment-related toxicity and mortality, raising the need to identify novel therapeutic approaches. High-throughput (HTP) drug screening enables the profiling of patients' responses in vitro and allows the repurposing of compounds currently used for other diseases, which can be immediately available for clinical application. The aim of this study was to apply HTP drug screening to identify potentially effective compounds for the treatment of pediatric BCP-ALL patients with poor prognosis, such as patients with Down Syndrome (DS) or carrying rearrangements involving PAX5 or KMT2A/MLL genes. Patient-derived Xenografts (PDX) samples from 34 BCP-ALL patients (9 DS CRLF2r, 15 PAX5r, 10 MLLr), 7 human BCP-ALL cell lines and 14 hematopoietic healthy donor samples were screened on a semi-automated HTP drug screening platform using a 174 compound library (FDA/EMA-approved or in preclinical studies). We identified 9 compounds active against BCP-ALL (ABT-199/venetoclax, AUY922/luminespib, dexamethasone, EC144, JQ1, NVP-HSP990, paclitaxel, PF-04929113 and vincristine), but sparing normal cells. Ex vivo validations confirmed that the BCL2 inhibitor venetoclax exerts an anti-leukemic effect against all three ALL subgroups at nanomolar concentrations. Overall, this study points out the benefit of HTP screening application for drug repurposing to allow the identification of effective and clinically translatable therapeutic agents for difficult-to-treat childhood BCP-ALL subgroups.
Collapse
Affiliation(s)
| | - Luigia Valsecchi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Manuel Quadri
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Titus Watrin
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Katerina Scharov
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Simona Procopio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Jia-Wey Tu
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Melina Vogt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Angela Maria Savino
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Daniela Silvestri
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Maria Grazia Valsecchi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy; Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Andrea Biondi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy; Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Giovanni Cazzaniga
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, Italy.
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Michela Bardini
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Chiara Palmi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
2
|
Zhang S, Zhou D, Zheng C, Xiong P, Zhu W, Zheng D. Preclinical evaluation of a novel antibody-drug conjugate targeting DR5 for lymphoblastic leukemia therapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 21:329-339. [PMID: 34141870 PMCID: PMC8173093 DOI: 10.1016/j.omto.2021.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/25/2021] [Indexed: 11/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological neoplasm resulting from immature lymphoid precursors. An antibody-drug conjugate (ADC), coupling a small molecule covalently with a targeting antibody, can specifically kill tumor cells. Death receptor 5 (DR5) is considered as a promising anti-tumor drug target. In this study, we describe the preclinical evaluation of a novel DR5-targeting ADC (Oba01) as a potential therapeutic against ALL. Oba01 utilizes anti-DR5 humanized monoclonal antibody (zaptuzumab) coupled via a cleavable linker to monomethyl auristatin E (MMAE). Oba01 can specifically bind to DR5 on the tumor cells and transfer into lysosome via DR5-mediated endocytosis. It then effectively releases the MMAE, which can bind to the tubulin and prevent its aggregation, thereby leading to a significant inhibition of proliferation and cell death in tumor cells. Additionally, Oba01 displays significant dose-dependent tumoricidal activity in cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. More importantly, toxicity analysis of Oba01 showed a favorable safety profile, and pharmacokinetic analysis illustrated an excellent stability and tolerability in rats and cynomolgus monkeys. Taken together, our data conclusively demonstrate that Oba01 is an attractive candidate for further clinical trials in DR5-positive ALL patients.
Collapse
Affiliation(s)
- Shuyong Zhang
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China.,Obio Technology (Shanghai) Corp, Ltd., No. 908, Building 19, Ziping Road, Pudong New District, Shanghai 201321, China
| | - Dongdong Zhou
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China
| | - Chao Zheng
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China
| | - Peng Xiong
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China
| | - Wan Zhu
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China
| | - Dexian Zheng
- Yantai Obioadc Biomedical Technology Ltd., Yantai, China.,Obio Technology (Shanghai) Corp, Ltd., No. 908, Building 19, Ziping Road, Pudong New District, Shanghai 201321, China
| |
Collapse
|
3
|
Palmi C, Savino AM, Silvestri D, Bronzini I, Cario G, Paganin M, Buldini B, Galbiati M, Muckenthaler MU, Bugarin C, Della Mina P, Nagel S, Barisone E, Casale F, Locatelli F, Lo Nigro L, Micalizzi C, Parasole R, Pession A, Putti MC, Santoro N, Testi AM, Ziino O, Kulozik AE, Zimmermann M, Schrappe M, Villa A, Gaipa G, Basso G, Biondi A, Valsecchi MG, Stanulla M, Conter V, Te Kronnie G, Cazzaniga G. CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:59260-59272. [PMID: 27449287 PMCID: PMC5312310 DOI: 10.18632/oncotarget.10610] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/01/2016] [Indexed: 12/01/2022] Open
Abstract
Pediatric T-ALL patients have a worse outcome compared to BCP-ALL patients and they could benefit from new prognostic marker identification. Alteration of CRLF2 gene, a hallmark correlated with poor outcome in BCP-ALL, has not been reported in T-ALL. We analyzed CRLF2 expression in 212 T-ALL pediatric patients enrolled in AIEOP-BFM ALL2000 study in Italian and German centers. Seventeen out of 120 (14.2%) Italian patients presented CRLF2 mRNA expression 5 times higher than the median (CRLF2-high); they had a significantly inferior event-free survival (41.2%±11.9 vs. 68.9%±4.6, p=0.006) and overall survival (47.1%±12.1 vs. 73.8%±4.3, p=0.009) and an increased cumulative incidence of relapse/resistance (52.9%±12.1 vs. 26.2%±4.3, p=0.007) compared to CRLF2-low patients. The prognostic value of CRLF2 over-expression was validated in the German cohort. Of note, CRLF2 over-expression was associated with poor prognosis in the high risk (HR) subgroup where CRLF2-high patients were more frequently allocated. Interestingly, although in T-ALL CRLF2 protein was localized mainly in the cytoplasm, in CRLF2-high blasts we found a trend towards a stronger TSLP-induced pSTAT5 response, sensitive to the JAK inhibitor Ruxolitinib. In conclusion, CRLF2 over-expression is a poor prognostic marker identifying a subset of HR T-ALL patients that could benefit from alternative therapy, potentially targeting the CRLF2 pathway.
Collapse
Affiliation(s)
- Chiara Palmi
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Angela M Savino
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Daniela Silvestri
- Center of Biostatistics for Clinical Epidemiology, Department of Health Sciences, University of Milano-Bicocca, Milan, Italy.,Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Ilaria Bronzini
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Gunnar Cario
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maddalena Paganin
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Barbara Buldini
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Marta Galbiati
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg and EMBL/Medical Faculty Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Cristina Bugarin
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Pamela Della Mina
- Microscopy and Image Analysis Consortium, Università di Milano-Bicocca, Monza, Italy
| | - Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Elena Barisone
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy
| | - Fiorina Casale
- Pediatric Oncology Service, Pediatric Department of 2nd University of Naples, Naples, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Ospedale Bambino Gesù, Rome - University of Pavia, Pavia, Italy
| | - Luca Lo Nigro
- Center of Pediatric Hematology Oncology, Azienda Ospedaliero-Universitaria "Policlinico Vittorio Emanuele", Catania, Italy
| | | | - Rosanna Parasole
- Department of Pediatric Hemato-Oncology, Ospedale Pausilipon, Napoli, Italy
| | - Andrea Pession
- Department of Pediatrics, "Lalla Seràgnoli" Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Maria C Putti
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Nicola Santoro
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University "A. Moro" of Bari, Bari, Italy
| | - Anna M Testi
- Division of Hematology, Department of Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy
| | - Ottavio Ziino
- Pediatric Hematology and Oncology Unit, A.R.N.A.S. Civico, Di Cristina and Benfratelli Hospital, Palermo, Italy
| | - Andreas E Kulozik
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg and EMBL/Medical Faculty Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Martin Zimmermann
- Department of Paediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Martin Schrappe
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Antonello Villa
- Microscopy and Image Analysis Consortium, Università di Milano-Bicocca, Monza, Italy
| | - Giuseppe Gaipa
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Giuseppe Basso
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Andrea Biondi
- Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Maria G Valsecchi
- Center of Biostatistics for Clinical Epidemiology, Department of Health Sciences, University of Milano-Bicocca, Milan, Italy
| | - Martin Stanulla
- Department of Paediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Valentino Conter
- Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Geertruy Te Kronnie
- Laboratory of Onco-Hematology, Department SDB, Università di Padova, Padova, Italy
| | - Giovanni Cazzaniga
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| |
Collapse
|
4
|
Forero-Castro M, Robledo C, Benito R, Bodega-Mayor I, Rapado I, Hernández-Sánchez M, Abáigar M, Maria Hernández-Sánchez J, Quijada-Álamo M, María Sánchez-Pina J, Sala-Valdés M, Araujo-Silva F, Kohlmann A, Luis Fuster J, Arefi M, de Las Heras N, Riesco S, Rodríguez JN, Hermosín L, Ribera J, Camos Guijosa M, Ramírez M, de Heredia Rubio CD, Barragán E, Martínez J, Ribera JM, Fernández-Ruiz E, Hernández-Rivas JM. Mutations in TP53 and JAK2 are independent prognostic biomarkers in B-cell precursor acute lymphoblastic leukaemia. Br J Cancer 2017; 117:256-265. [PMID: 28557976 PMCID: PMC5520505 DOI: 10.1038/bjc.2017.152] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In B-cell precursor acute lymphoblastic leukaemia (B-ALL), the identification of additional genetic alterations associated with poor prognosis is still of importance. We determined the frequency and prognostic impact of somatic mutations in children and adult cases with B-ALL treated with Spanish PETHEMA and SEHOP protocols. METHODS Mutational status of hotspot regions of TP53, JAK2, PAX5, LEF1, CRLF2 and IL7R genes was determined by next-generation deep sequencing in 340 B-ALL patients (211 children and 129 adults). The associations between mutation status and clinicopathological features at the time of diagnosis, treatment outcome and survival were assessed. Univariate and multivariate survival analyses were performed to identify independent prognostic factors associated with overall survival (OS), event-free survival (EFS) and relapse rate (RR). RESULTS A mutation rate of 12.4% was identified. The frequency of adult mutations was higher (20.2% vs 7.6%, P=0.001). TP53 was the most frequently mutated gene (4.1%), followed by JAK2 (3.8%), CRLF2 (2.9%), PAX5 (2.4%), LEF1 (0.6%) and IL7R (0.3%). All mutations were observed in B-ALL without ETV6-RUNX1 (P=0.047) or BCR-ABL1 fusions (P<0.0001). In children, TP53mut was associated with lower OS (5-year OS: 50% vs 86%, P=0.002) and EFS rates (5-year EFS: 50% vs 78.3%, P=0.009) and higher RR (5-year RR: 33.3% vs 18.6% P=0.037), and was independently associated with higher RR (hazard ratio (HR)=4.5; P=0.04). In adults, TP53mut was associated with a lower OS (5-year OS: 0% vs 43.3%, P=0.019) and a higher RR (5-year RR: 100% vs 61.4%, P=0.029), whereas JAK2mut was associated with a lower EFS (5-year EFS: 0% vs 30.6%, P=0.035) and a higher RR (5-year RR: 100% vs 60.4%, P=0.002). TP53mut was an independent risk factor for shorter OS (HR=2.3; P=0.035) and, together with JAK2mut, also were independent markers of poor prognosis for RR (TP53mut: HR=5.9; P=0.027 and JAK2mut: HR=5.6; P=0.036). CONCLUSIONS TP53mut and JAK2mut are potential biomarkers associated with poor prognosis in B-ALL patients.
Collapse
Affiliation(s)
- Maribel Forero-Castro
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain.,School of Biological Sciences (GICBUPTC research group), Universidad Pedagógica y Tecnológica de Colombia (UPTC), Avenida Central del Norte 39-115, Tunja 150003, Colombia
| | - Cristina Robledo
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Rocío Benito
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Irene Bodega-Mayor
- Molecular Biology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Calle Diego de León, 62, Madrid 28006, Spain
| | - Inmaculada Rapado
- Department of Hematology, Hospital 12 de Octubre, Avenida de Córdoba s/n, Madrid 28041, Spain
| | - María Hernández-Sánchez
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - María Abáigar
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Jesús Maria Hernández-Sánchez
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Miguel Quijada-Álamo
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - José María Sánchez-Pina
- Department of Hematology, Hospital 12 de Octubre, Avenida de Córdoba s/n, Madrid 28041, Spain
| | - Mónica Sala-Valdés
- Molecular Biology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Calle Diego de León, 62, Madrid 28006, Spain
| | - Fernanda Araujo-Silva
- Molecular Biology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Calle Diego de León, 62, Madrid 28006, Spain
| | - Alexander Kohlmann
- Personalised Healthcare and Biomarkers, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Darwin Building, 310 Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - José Luis Fuster
- Department of Pediatric Oncohematology, Hospital Universitario Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, El Palmar, Murcia 30120, Spain
| | - Maryam Arefi
- Department of Hematology, Hospital Río Carrión, Av. Donantes de Sangre, s/n, Palencia 34005, Spain
| | - Natalia de Las Heras
- Department of Hematology, Hospital Virgen Blanca, Altos de Nava s/n, León 24071, Spain
| | - Susana Riesco
- Department of Pediatrics, Hospital Universitario de Salamanca, Paseo de San Vicente, 88-182, Salamanca 37007, Spain
| | - Juan N Rodríguez
- Department of Hematology, Hospital Juan Ramón Jiménez, Ronda Exterior Norte, s/n, Huelva 21005, Spain
| | - Lourdes Hermosín
- Department of Hematology, Hospital de Jerez, Carr Madrid-Cádiz, Jerez de la Frontera 11407, Cádiz, Spain
| | - Jordi Ribera
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Instituto de Investigación Josep Carreras, (Can Ruti), Carretera de Canyet, s/n, Badalona, Barcelona 08916, Spain
| | - Mireia Camos Guijosa
- Hematology Laboratory, Institut de Recerca Pediátrica Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu, 2, Esplugues de Llobregat, Barcelona 08950, Spain
| | - Manuel Ramírez
- Pediatric Oncohematology, Hospital Universitario Infantil Niño Jesús, Instituto de Investigación Sanitaria Princesa (IIS-IP), Av. de Menéndez Pelayo, 65, Madrid 28009, Spain
| | | | - Eva Barragán
- Molecular Biology Lab, Clinical Analysis Service, Hospital Universitario y Politécnico de La Fe, Avinguda de Fernando Abril Martorell, 106, Valencia 46026, Spain
| | - Joaquín Martínez
- Department of Hematology, Hospital 12 de Octubre, Avenida de Córdoba s/n, Madrid 28041, Spain
| | - José M Ribera
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Instituto de Investigación Josep Carreras, (Can Ruti), Carretera de Canyet, s/n, Badalona, Barcelona 08916, Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Calle Diego de León, 62, Madrid 28006, Spain
| | - Jesús-María Hernández-Rivas
- IBSAL, IBMCC, University of Salamanca, CSIC, Cancer Research Center, Campus Miguel de Unamuno, Salamanca 37007, Spain.,Department of Hematology, Hospital Universitario de Salamanca, Paseo de San Vicente, 88-182, Salamanca 37007, Spain
| |
Collapse
|
5
|
BCR-ABL (Ph)-like acute leukemia—Pathogenesis, diagnosis and therapeutic options. Blood Rev 2017; 31:11-16. [DOI: 10.1016/j.blre.2016.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 08/23/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022]
|
6
|
Bartram J, Wade R, Vora A, Hancock J, Mitchell C, Kinsey S, Steward C, Moppett J, Goulden N. Excellent outcome of minimal residual disease-defined low-risk patients is sustained with more than 10 years follow-up: results of UK paediatric acute lymphoblastic leukaemia trials 1997-2003. Arch Dis Child 2016; 101:449-54. [PMID: 26865705 DOI: 10.1136/archdischild-2015-309617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/20/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) is defined as the presence of sub-microscopic levels of leukaemia. Measurement of MRD from bone marrow at the end of induction chemotherapy (day 28) for childhood acute lymphoblastic leukaemia (ALL) can highlight a large group of patients (>40%) with an excellent (>90%) short-term event-free survival (EFS). However, follow-up in recent published trials is relatively short, raising concerns about using this result to infer the safety of further therapy reduction in the future. METHODS We examined MRD data on 225 patients treated on one of three UKALL trials between 1997 and 2003 to assess the long-term (>10 years follow-up) outcome of those patients who had low-risk MRD (<0.01%) at day 28. RESULTS Our pilot data define a cohort of 53% of children with MRD <0.01% at day 28 who have an EFS of 91% and long-term overall survival of 97%. Of 120 patients with day-28 MRD <0.01% and extended follow-up, there was one death due to treatment-related toxicity, one infectious death while in complete remission, and four relapse deaths. CONCLUSIONS The excellent outcome for childhood ALL in patients with MRD <0.01% after induction chemotherapy is sustained for more than 10 years from diagnosis. This supports the potential exploration of further reduction of therapy in this group, in an attempt to reduce treatment-related mortality and late effects.
Collapse
Affiliation(s)
- Jack Bartram
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Rachel Wade
- Clinical Trial Service Unit, University of Oxford, Oxford, UK
| | - Ajay Vora
- Department of Haematology, Sheffield Children's Hospital, Sheffield, UK
| | - Jeremy Hancock
- Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Chris Mitchell
- Paediatric Haematology and Oncology, John Radcliffe Hospital, Oxford, UK
| | - Sally Kinsey
- Department of Paediatric Haematology, St James' University Hospital, Leeds, UK
| | - Colin Steward
- Department of Paediatric Haematology/Oncology, Royal Hospital for Children, Bristol, UK
| | - John Moppett
- Department of Paediatric Haematology/Oncology, Royal Hospital for Children, Bristol, UK
| | - Nick Goulden
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
7
|
Chen C, Bartenhagen C, Gombert M, Okpanyi V, Binder V, Röttgers S, Bradtke J, Teigler-Schlegel A, Harbott J, Ginzel S, Thiele R, Husemann P, Krell PF, Borkhardt A, Dugas M, Hu J, Fischer U. Next-generation-sequencing of recurrent childhood high hyperdiploid acute lymphoblastic leukemia reveals mutations typically associated with high risk patients. Leuk Res 2015; 39:990-1001. [DOI: 10.1016/j.leukres.2015.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 01/07/2023]
|
8
|
Hakeem A, Shiekh AA, Bhat GM, Lone AR. Prognostification of ALL by Cytogenetics. Indian J Hematol Blood Transfus 2015; 31:322-31. [PMID: 26085716 PMCID: PMC4465518 DOI: 10.1007/s12288-014-0483-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/20/2014] [Indexed: 10/24/2022] Open
Abstract
Cytogenetic abnormalities in chromosomal number and structure are common in pediatric ALL and some have prognostic significance. One interesting association between cytogenetic status and treatment response involves the metabolism of methotrexate. Hyperdiploid lymphoblasts accumulate increased amounts of MTX and MTX polyglutamates, and they have higher basal apoptotic rates compared with leukemic cells with lower ploidy and normal cells. These characteristics may contribute to the better outcomes observed for patients with hyperdiploid lymphoblasts. A number of recurrent chromosomal abnormalities have been shown to have prognostic significance, especially in B-precursor ALL. Some chromosomal abnormalities are associated with more favorable outcomes, such as high hyperdiploidy (51-65 chromosomes) and the ETV6-RUNX1 fusion. Others are associated with a poorer prognosis, including the Philadelphia chromosome [t(9;22)], rearrangements of the MLL gene (chromosome 11q23), and intrachromosomal amplification of the AML1 gene (iAMP21).
Collapse
Affiliation(s)
- Ansar Hakeem
- Department of Med Oncology, SKIMS SGR, Srinagar, 190011 J And K India
| | - Aejaz Aziz Shiekh
- Department of Med Oncology, SKIMS SGR, Srinagar, 190011 J And K India
| | - Gull Mohd. Bhat
- Department of Med Oncology, SKIMS SGR, Srinagar, 190011 J And K India
| | - A. R. Lone
- Department of Med Oncology, SKIMS SGR, Srinagar, 190011 J And K India
| |
Collapse
|
9
|
Dixon-McIver A. Emerging technologies in paediatric leukaemia. Transl Pediatr 2015; 4:116-24. [PMID: 26835367 PMCID: PMC4729090 DOI: 10.3978/j.issn.2224-4336.2015.03.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Genetic changes, in particular chromosomal aberrations, are a hallmark of acute lymphoblastic lymphoma (ALL) and accurate detection of them is important in ensuring assignment to the appropriate drug protocol. Our ability to detect these genetic changes has been somewhat limited in the past due to the necessity to analyse mitotically active cells by conventional G-banded metaphase analysis and by mutational analysis of individual genes. Advances in technology include high resolution, microarray-based techniques that permit examination of the whole genome. Here we will review the current available methodology and discuss how the technology is being integrated into the diagnostic setting.
Collapse
|
10
|
Gabriel AS, Lafta FM, Schwalbe EC, Nakjang S, Cockell SJ, Iliasova A, Enshaei A, Schwab C, Rand V, Clifford SC, Kinsey SE, Mitchell CD, Vora A, Harrison CJ, Moorman AV, Strathdee G. Epigenetic landscape correlates with genetic subtype but does not predict outcome in childhood acute lymphoblastic leukemia. Epigenetics 2015; 10:717-26. [PMID: 26237075 PMCID: PMC4622588 DOI: 10.1080/15592294.2015.1061174] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 05/29/2015] [Accepted: 06/05/2015] [Indexed: 11/19/2022] Open
Abstract
Although children with acute lymphoblastic leukemia (ALL) generally have a good outcome, some patients do relapse and survival following relapse is poor. Altered DNA methylation is highly prevalent in ALL and raises the possibility that DNA methylation-based biomarkers could predict patient outcome. In this study, genome-wide methylation analysis, using the Illumina Infinium HumanMethylation450 BeadChip platform, was carried out on 52 diagnostic patient samples from 4 genetic subtypes [ETV6-RUNX1, high hyperdiploidy (HeH), TCF3-PBX1 and dic(9;20)(p11-13;q11)] in a 1:1 case-control design with patients who went on to relapse (as cases) and patients achieving long-term remission (as controls). Pyrosequencing assays for selected loci were used to confirm the array-generated data. Non-negative matrix factorization consensus clustering readily clustered samples according to genetic subgroups and gene enrichment pathway analysis suggested that this is in part driven by epigenetic disruption of subtype specific signaling pathways. Multiple bioinformatics approaches (including bump hunting and individual locus analysis) were used to identify CpG sites or regions associated with outcome. However, no associations with relapse were identified. Our data revealed that ETV6-RUNX1 and dic(9;20) subtypes were mostly associated with hypermethylation; conversely, TCF3-PBX1 and HeH were associated with hypomethylation. We observed significant enrichment of the neuroactive ligand-receptor interaction pathway in TCF3-PBX1 as well as an enrichment of genes involved in immunity and infection pathways in ETV6-RUNX1 subtype. Taken together, our results suggest that altered DNA methylation may have differential impacts in distinct ALL genetic subtypes.
Collapse
Affiliation(s)
- Alem S Gabriel
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Fadhel M Lafta
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Edward C Schwalbe
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
- Department of Applied Sciences; Northumbria University; Newcastle upon Tyne, UK
| | - Sirintra Nakjang
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
- Bioinformatics Support Unit; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Simon J Cockell
- Bioinformatics Support Unit; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Alice Iliasova
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
- Department of Applied Sciences; Northumbria University; Newcastle upon Tyne, UK
| | - Amir Enshaei
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Claire Schwab
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Vikki Rand
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Steven C Clifford
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Sally E Kinsey
- Department of Pediatric Haematology and Oncology; Leeds General Infirmary; Leeds, UK
| | - Chris D Mitchell
- Department of Pediatric Oncology; John Radcliffe Hospital; Oxford, UK
| | - Ajay Vora
- Department of Pediatric Oncology; John Radcliffe Hospital; Oxford, UK
| | - Christine J Harrison
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Anthony V Moorman
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
| | - Gordon Strathdee
- Northern Institute for Cancer Research; Faculty of Medical Sciences; Newcastle University; Newcastle upon Tyne, UK
- Department of Haematology; Great Ormond Street Hospital; London, UK
| |
Collapse
|
11
|
Ceppi F, Cazzaniga G, Colombini A, Biondi A, Conter V. Risk factors for relapse in childhood acute lymphoblastic leukemia: prediction and prevention. Expert Rev Hematol 2014; 8:57-70. [PMID: 25367188 DOI: 10.1586/17474086.2015.978281] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
With current treatment regimens, survival rates for acute lymphoblastic leukemia (ALL) have improved dramatically since the 1980s, with current 5-year overall survival rates estimated at greater than 85%. This success was achieved, in part, through the implementation of risk-stratified therapy. Nevertheless, for a subgroup of patients (15-20%) with newly diagnosed ALL who will ultimately relapse, traditional risk assessment remains inadequate. The risk of relapse may be estimated on the basis of diagnostic features or early treatment response findings. Further progress in this field may thus come from refinement of predictive factors for relapse and treatment adaptation and from the identification of biological subsets of ALL patients who could benefit from specific target therapies. This article summarizes the aspects associated with the identification of predictive factors for relapse in childhood ALL and options available for prevention of disease recurrence.
Collapse
Affiliation(s)
- Francesco Ceppi
- Division of Haematology/Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
12
|
Vora A, Goulden N, Mitchell C, Hancock J, Hough R, Rowntree C, Moorman AV, Wade R. Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): a randomised controlled trial. Lancet Oncol 2014; 15:809-18. [PMID: 24924991 DOI: 10.1016/s1470-2045(14)70243-8] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND No randomised study has shown whether stratification of treatment by minimal residual disease (MRD) response improves outcome in children and young people with acute lymphoblastic leukaemia (ALL). We assessed whether children and young people with clinical standard and intermediate-risk ALL who have persistent MRD at the end of induction therapy benefit from augmented post-remission therapy. METHODS Between Oct 1, 2003, and June 30, 2011, we enrolled eligible patients aged 1-24 years and initially categorised them into clinical standard-risk, intermediate-risk, and high-risk groups on the basis of a combination of National Cancer Institute criteria, cytogenetics, and early morphological response to induction therapy. Clinical standard-risk and intermediate-risk patients with MRD of 0·01% or higher at day 29 of induction (MRD high risk) were randomly assigned (1:1) to standard therapy (treatment regimens A and B) or augmented post-remission therapy (regimen C). Compared with standard therapy, the augmented treatment regimen (regimen C) included an additional eight doses of pegylated asparaginase, 18 doses of vincristine, and escalated-dose intravenous methotrexate without folinic acid rescue during interim maintenance courses. Computer randomisation was used for treatment allocation and was balanced for sex, age (<10 years vs ≥10 years), and white blood cell count at diagnosis (<50 × 10(9)/L vs ≥50 × 10(9)/L) by minimisation. Patients, clinicians, and data analysts were not masked to treatment allocation. The primary outcomes were event-free survival and overall survival. Analyses were by intention to treat. This trial is registered with Current Controlled Trials, number ISRCTN07355119. FINDINGS 533 MRD high-risk patients were randomly assigned to receive standard (n=266) or augmented (n=267) post-remission therapy. After a median follow-up of 70 months (IQR 52-91), 5-year event-free survival was better in the augmented treatment group (89·6% [95% CI 85·9-93·3]) than in the standard group (82·8% [78·1-87·5]; odds ratio [OR] 0·61 [95% CI 0·39-0·98], p=0·04). Overall survival at 5 years was numerically, but not significantly, higher in the augmented treatment group (92·9% [95% CI 89·8-96·0]) than in the standard therapy group (88·9% [85·0-92·8]; OR 0·67 [95% CI 0·38-1·17], p=0·16). More adverse events occurred in the augmented treatment group than in the standard group (asparaginase-related hypersensitivity in 18 [6·7%] in the augmented group vs two [0·8%] in the standard group and asparaginase-related pancreatitis in eight [3·0%] vs one [0·4%]; intravenous methotrexate-related mucositis in 11 [4·1%] vs three [1·1%] and methotrexate-related stomatitis in 48 [18·0%] vs 12 [4·5%]). INTERPRETATION Our findings suggest that children and young people with acute lymphoblastic leukaemia and 0·01% or more MRD at the end of remission induction therapy could benefit from augmented post-remission therapy. However, the asparaginase and intravenous methotrexate used in the augmented treatment regimen is associated with more adverse events than is the standard post-remission treatment regimen. FUNDING Medical Research Council and Leukaemia and Lymphoma Research.
Collapse
Affiliation(s)
- Ajay Vora
- Sheffield Children's Hospital, Sheffield, UK.
| | | | | | | | | | | | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Northern Institute for Cancer Research, Newcastle University, Newcastle, UK
| | - Rachel Wade
- Clinical Trial Service Unit, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Juárez-Velázquez R, Reyes-León A, Salas-Labadía C, Rivera-Luna R, Velasco-Hidalgo L, López-Hernández G, López-Santiago N, Paredes-Aguilera R, Domínguez-López A, Bernáldez R, Pérez-Vera P. Significance of CASP8AP2 and H2AFZ expression in survival and risk of relapse in children with acute lymphoblastic leukemia. Leuk Lymphoma 2014; 55:2305-11. [PMID: 24397596 DOI: 10.3109/10428194.2013.878458] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Novel biomarkers for risk refinement and stratification in childhood acute lymphoblastic leukemia (ALL) are needed to optimize treatment results. We studied the expression of CASP8AP2 and H2AFZ associated with relapse and survival in bone marrow samples from newly diagnosed children with ALL. We found: (a) an increased risk for early relapse in those patients with low expression of CASP8AP2 (odds ratio [OR] 3.93, 95% confidence interval [CI] 1.40-11.02, p < 0.05) confirming its usefulness as a predictive risk marker, although H2AFZ did not present the same effect; (b) patients with low expressions of CASP8AP2 and H2AFZ had inferior survival rates (p < 0.001); (c) the predictive values regarding low expressions of H2AFZ and CASP8AP2 and high white blood cell count suggest that these features could help to identify more accurately patients at greater risk of relapse.
Collapse
Affiliation(s)
- Rocío Juárez-Velázquez
- Laboratorio de Cultivo de Tejidos, Instituto Nacional de Pediatría , México D.F. , México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Izraeli S, Shochat C, Tal N, Geron I. Towards precision medicine in childhood leukemia--insights from mutationally activated cytokine receptor pathways in acute lymphoblastic leukemia. Cancer Lett 2014; 352:15-20. [PMID: 24569093 DOI: 10.1016/j.canlet.2014.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 02/08/2014] [Accepted: 02/10/2014] [Indexed: 01/30/2023]
Abstract
The successful therapy of childhood leukemia has been characterized by careful personalized adaptation of therapy by risk stratification. Yet almost all drugs are relatively non-specific. To achieve greater precision in therapy, druggable targets and specific targeting drugs are necessary. Here we review the recent discoveries of cytokine receptors and their signaling components in high risk leukemias and the potential approaches to target them.
Collapse
Affiliation(s)
- Shai Izraeli
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel.
| | - Chen Shochat
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel; Migal Galilee Technology Center, Kiryat Shmona, Israel; Tel Hai College, Upper Galilee 12210, Israel
| | - Noa Tal
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Ifat Geron
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel; Division of Biological Sciences and Department of Medicine Stem Cell Program, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
15
|
Abstract
Abstract
Acute lymphoblastic leukemia in childhood has shown remarkable improvements in outcome over the past decades. This achievement was the result of better patient risk assessment, intensification of treatment, appropriate use of BM transplantation, and improved supportive therapies. Among risk factors, early response (originally morphologic and today minimal residual disease) has acquired a prominent role. The predictive value of minimal residual disease evaluation as a measurement of in vivo drug resistance opened new perspectives for its use in clinical evaluation to determine a risk-based treatment and as a potential surrogate end point for efficacy. More recently, detailed genomic analyses of childhood acute lymphoblastic leukemia have increased our knowledge in this disease. It is likely that this will lead to further improvement of risk assessment and stratification to targeted therapies. Leukemic subsets defined on the basis of biological mechanisms and driver mutations will be ever smaller. To facilitate continued progress, this new scenario will raise methodological issues in study design and the need for collaboration across large, well-characterized patient populations.
Collapse
|
16
|
Wade CH, Tarini BA, Wilfond BS. Growing up in the genomic era: implications of whole-genome sequencing for children, families, and pediatric practice. Annu Rev Genomics Hum Genet 2013; 14:535-55. [PMID: 23875800 DOI: 10.1146/annurev-genom-091212-153425] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Whole-genome sequencing (WGS) has advanced to a point where it is beginning to be integrated into pediatric practice. With little consensus on how to maximize the benefits of WGS for children, there is a growing need for focused efforts that connect researchers, clinicians, and families to chart a path forward. To illustrate relevant concerns, two contrasting applications of pediatric WGS are explored: clinical use with children who have undiagnosed conditions, and population-based screening. Specific challenges for health care services, policy development, and the well-being of children are discussed in light of current research. In the interest of ensuring evidence-based pediatric WGS, strategies are identified for advancing our understanding of what it means for children to grow up with WGS results guiding their health care.
Collapse
Affiliation(s)
- Christopher H Wade
- Nursing and Health Studies Program, University of Washington Bothell, Bothell, Washington 98011;
| | | | | |
Collapse
|
17
|
Vora A, Goulden N, Wade R, Mitchell C, Hancock J, Hough R, Rowntree C, Richards S. Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial. Lancet Oncol 2013; 14:199-209. [DOI: 10.1016/s1470-2045(12)70600-9] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
18
|
Moorman AV. The clinical relevance of chromosomal and genomic abnormalities in B-cell precursor acute lymphoblastic leukaemia. Blood Rev 2012; 26:123-35. [DOI: 10.1016/j.blre.2012.01.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Poor prognosis for P2RY8-CRLF2 fusion but not for CRLF2 over-expression in children with intermediate risk B-cell precursor acute lymphoblastic leukemia. Leukemia 2012; 26:2245-53. [PMID: 22484421 DOI: 10.1038/leu.2012.101] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL) has achieved an 80% cure rate as a result of a risk-adapted therapy largely based on minimal residual disease (MRD) monitoring. However, relapse is still the most frequent adverse event, occurring mainly in the patients with intermediate MRD levels (intermediate risk, IR), emphasizing the need for new prognostic markers. We analyzed the prognostic impact of cytokine receptor-like factor 2 (CRLF2) over-expression and P2RY8-CRLF2 fusion in 464 BCP-ALL patients (not affected by Down syndrome and BCR-ABL negative) enrolled in the AIEOP-BFM ALL2000 study in Italy. In 22/464 (4.7%) samples, RQ-PCR showed CRLF2 over-expression (≥20 times higher than the overall median). P2RY8-CRLF2 fusion was detected in 22/365 (6%) cases, with 10/22 cases also showing CRLF2 over-expression. P2RY8-CRLF2 fusion was the most relevant prognostic factor independent of CRLF2 over-expression with a threefold increase in risk of relapse. Significantly, the cumulative incidence of relapse of the P2RY8-CRLF2 + patients in the IR group was high (61.1% ± 12.9 vs 17.6% ± 2.6, P<0.0001), similar to high-risk patients in AIEOP-BFM ALL2000 study. These results were confirmed in a cohort of patients treated in Germany. In conclusion, P2RY8-CRLF2 identifies a subset of BCP-ALL patients currently stratified as IR that could be considered for treatment intensification.
Collapse
|
20
|
Tang W, Hu Z, Muallem H, Gulley ML. Quality assurance of RNA expression profiling in clinical laboratories. J Mol Diagn 2012; 14:1-11. [PMID: 22020152 PMCID: PMC3338342 DOI: 10.1016/j.jmoldx.2011.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/09/2011] [Accepted: 09/14/2011] [Indexed: 12/12/2022] Open
Abstract
RNA expression profiles are increasingly used to diagnose and classify disease, based on expression patterns of as many as several thousand RNAs. To ensure quality of expression profiling services in clinical settings, a standard operating procedure incorporates multiple quality indicators and controls, beginning with preanalytic specimen preparation and proceeding thorough analysis, interpretation, and reporting. Before testing, histopathological examination of each cellular specimen, along with optional cell enrichment procedures, ensures adequacy of the input tissue. Other tactics include endogenous controls to evaluate adequacy of RNA and exogenous or spiked controls to evaluate run- and patient-specific performance of the test system, respectively. Unique aspects of quality assurance for array-based tests include controls for the pertinent outcome signatures that often supersede controls for each individual analyte, built-in redundancy for critical analytes or biochemical pathways, and software-supported scrutiny of abundant data by a laboratory physician who interprets the findings in a manner facilitating appropriate medical intervention. Access to high-quality reagents, instruments, and software from commercial sources promotes standardization and adoption in clinical settings, once an assay is vetted in validation studies as being analytically sound and clinically useful. Careful attention to the well-honed principles of laboratory medicine, along with guidance from government and professional groups on strategies to preserve RNA and manage large data sets, promotes clinical-grade assay performance.
Collapse
Affiliation(s)
- Weihua Tang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hind Muallem
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Margaret L. Gulley
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
21
|
Tang W, Hu Z, Muallem H, Gulley ML. Clinical implementation of RNA signatures for pharmacogenomic decision-making. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2011; 4:95-107. [PMID: 23226056 PMCID: PMC3513222 DOI: 10.2147/pgpm.s14888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Indexed: 12/14/2022]
Abstract
RNA profiling is increasingly used to predict drug response, dose, or toxicity based on analysis of drug pharmacokinetic or pharmacodynamic pathways. Before implementing multiplexed RNA arrays in clinical practice, validation studies are carried out to demonstrate sufficient evidence of analytic and clinical performance, and to establish an assay protocol with quality assurance measures. Pathologists assure quality by selecting input tissue and by interpreting results in the context of the input tissue as well as the technologies that were used and the clinical setting in which the test was ordered. A strength of RNA profiling is the array-based measurement of tens to thousands of RNAs at once, including redundant tests for critical analytes or pathways to promote confidence in test results. Instrument and reagent manufacturers are crucial for supplying reliable components of the test system. Strategies for quality assurance include careful attention to RNA preservation and quality checks at pertinent steps in the assay protocol, beginning with specimen collection and proceeding through the various phases of transport, processing, storage, analysis, interpretation, and reporting. Specimen quality is checked by probing housekeeping transcripts, while spiked and exogenous controls serve as a check on analytic performance of the test system. Software is required to manipulate abundant array data and present it for interpretation by a laboratory physician who reports results in a manner facilitating therapeutic decision-making. Maintenance of the assay requires periodic documentation of personnel competency and laboratory proficiency. These strategies are shepherding genomic arrays into clinical settings to provide added value to patients and to the larger health care system.
Collapse
Affiliation(s)
- Weihua Tang
- Department of Pathology and Laboratory Medicine
| | | | | | | |
Collapse
|
22
|
Harrison CJ, Haas O, Harbott J, Biondi A, Stanulla M, Trka J, Izraeli S. Detection of prognostically relevant genetic abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: recommendations from the Biology and Diagnosis Committee of the International Berlin-Frankfürt-Münster study group. Br J Haematol 2010; 151:132-42. [DOI: 10.1111/j.1365-2141.2010.08314.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|